Skip to main content
Endocrinology logoLink to Endocrinology
. 2011 Jun 28;152(9):3306–3311. doi: 10.1210/en.2011-1104

Minireview: Cracking the Metabolic Code for Thyroid Hormone Signaling

Antonio C Bianco 1,
PMCID: PMC3159779  PMID: 21712363

Abstract

Cells are not passive bystanders in the process of hormonal signaling and instead can actively customize hormonal action. Thyroid hormone gains access to the intracellular environment via membrane transporters, and while diffusing from the plasma membrane to the nucleus, thyroid hormone signaling is modified via the action of the deiodinases. Although the type 2 deiodinase (D2) converts the prohormone T4 to the biologically active T3, the type 3 deiodinase (D3) converts it to reverse T3, an inactive metabolite. D3 also inactivates T3 to T2, terminating thyroid hormone action. Therefore, D2 confers cells with the capacity to produce extra amounts of T3 and thus enhances thyroid hormone signaling. In contrast expression of D3 results in the opposite action. The Dio2 and Dio3 genes undergo transcriptional regulation throughout embryonic development, childhood, and adult life. In addition, the D2 protein is unique in that it can be switched off and on via an ubiquitin regulated mechanism, triggered by catalysis of T4. Induction of D2 enhances local thyroid hormone signaling and energy expenditure during activation of brown adipose tissue by cold exposure or high-fat diet. On the other hand, induction of D3 in myocardium and brain during ischemia and hypoxia decreases energy expenditure as part of a homeostatic mechanism to slow down cell metabolism in the face of limited O2 supply.


Most eukaryotic cells are equipped with built-in genetic programs that control cell division, homeostatic functions, and basic phenotype. These cellular programs can be modified by environmental cues, such as nutrient availability or biologically active molecules, the nature and intensity over which most cells have very little control. Hormones constitute an example of such biologically active molecules, and cells are generally classified as responsive or unresponsive, depending on whether they have sufficient number of their cognate receptors. However, it has become increasingly clear that cells are not passive bystanders in this process and instead can actively customize hormonal signals, such as with sexual steroids and thyroid hormone. For example, cellular expression of 5α-reductase or P450 aromatase, respectively, transforms the testosterone molecule into dihydrotestosterone or estradiol, locally changing testosterone's biological activity in opposite directions. A similar scenario exists in the case of the deiodinases, enzymes that can locally activate or inactivate thyroid hormone.

Cellular membranes are relatively impermeable to thyroid hormone and thus membrane transporters are necessary for access to the intracellular environment (1). Once inside the cells, thyroid hormone diffuses toward the nucleus and eventually binds to its receptors, high affinity ligand-dependent transcription factors that modify gene expression (2). However, inside the cells, the prohormone T4 can be transformed to the biologically active T3 molecule via the type 2 deiodinase (D2), or it can be inactivated to form reverse T3 via the type 3 deiodinase (D3). Most importantly, T3 is also inactivated by D3, preventing or terminating thyroid hormone action (3). Thus, although diffusing from the plasma membrane to the nucleus, thyroid hormone signaling is modified via the action of the deiodinases.

Deiodinases are dimeric integral-membrane thyroredoxin fold-containing selenoproteins of about 60 kDa (dimer) (49). Each dimer counterpart consists of a selenocystein-containing globular domain that is anchored to cellular membranes through a single amino-terminal transmembrane segment. D2 is an endoplasmic reticulum (ER)-resident protein that is retained in ER and generates T3 in the proximity of the nuclear compartment (10). On the other hand, most D3 goes through the Golgi complex and reaches the plasma membrane, where it undergoes endocytosis and recycles via the early endosomes (4). Thus, D2 expression confers cells with the capacity to produce additional amounts of T3 and thus enhances thyroid hormone signaling. In contrast, expression of D3 results in the opposite action. Furthermore, these events occur in the cell without relative changes to plasma thyroid hormone levels (11, 12).

The Dio2 and Dio3 genes undergo transcriptional regulation throughout embryonic development, childhood, and adult life (11). Dio2 is a highly sensitive cAMP-responsive gene (13) that is also positively regulated by nuclear factor κB (14) and forkhead box (Fox)O3 (15). At the same time, Dio3 is up-regulated by retinoic acid, 12-O-tetradecanoyl phorbol 13-acetate, basic fibroblast growth factor (16), TGFβ (17), the hedgehog-GLI family zinc finger 2 pathway (18), and hypoxia-inducible factor-1α (19).

The D2 protein is unique in that it can be switched off and on via an ubiquitin (Ub)-regulated mechanism, triggered by catalysis of T4 (2022). It is assumed that T4 deiodination exposes Lys-residues in D2's globular domain that are subsequently conjugated to Ub. Moreover, this results in inactivating D2 by disruption of the dimer formation (20). Ub-D2 is not immediately taken up by the proteasome and instead can be deubiquitinated and reactivated to produce another molecule of T3, repeating the cycle. Although two Ub conjugases are involved in the process of D2 ubiquitination (23, 24), the limiting components of this pathway are two E3-ligase adaptors. These include the hedgehog-inducible suppressor of cytokine signaling-box containing WD repeat suppressor of cytokine signaling box-containing protein-1 (25) and TEB4 (26), a ligase involved in the ER-associated degradation program. In contrast, two Ub-specific proteases (USP), USP20 and USP33, mediate deubiquitination and reactivation of Ub-D2 (21).

Thus, it is clear that thyroid hormone levels in the plasma do not faithfully reflect thyroid hormone signaling in cells; this action takes place inside the cell. A complex network of transcriptional and posttranscriptional mechanisms regulating deiodinase expression is at work in health and disease, mediating rapid customization of thyroid hormone signaling on a cell-specific basis.

Deiodinases and the Metabolic Effects of Thyroid Hormone

Insulin typifies how metabolic pathways are controlled by systemic hormones. In the minutes that follow a meal, insulin is secreted into the bloodstream, exposing all tissues to elevated levels of insulin. As a result, glucose uptake and oxidation is increased, the synthesis of fatty acid is accelerated, and protein anabolism is enhanced. A few hours later, plasma insulin levels are back to premeal levels and so are its metabolic effects. This is a very different model to that which cells respond to thyroid hormone. On the contrary, thyroid hormone levels in the plasma hardly fluctuate in healthy individuals, as shown in a year-long study of serum levels of T4 and T3 (27). Thus, thyroid hormone-responsive metabolic processes are turned on and off by thyroid hormone via deiodination pathways that are taking place inside the target cells, seemingly invisible from the plasma viewpoint (11).

Even though we are just starting to understand these pathways, some thyroid hormone effects on metabolism are well recognized, including acceleration of substrate cycles, ionic cycles, and mitochondrial respiration, all leading to accelerated energy expenditure (28). Unfortunately, most of what we know about these effects is from nonphysiological models in which subjects were systemically hypothyroid or thyrotoxic. To illustrate this point, hypothyroidism and thyrotoxicosis are known to affect sympathetic outflow to a number of metabolically active tissues, such as white and brown adipose tissue (BAT), liver, skeletal muscle, and heart (29, 30). These states mask the true effects of thyroid hormone deficiency or its excess. This is illustrated in the D2 knockout (D2KO) mice. At room temperature, which is considered a significant thermal stress for mice, D2KO mice preferentially oxidize fat, have a normal sensitivity to diet-induced obesity, and are supertolerant to glucose load. However, when thermal stress is eliminated and sympathetic activity minimized at thermoneutrality (30 C), an opposite phenotype is encountered, one that includes obesity, glucose intolerance, and exacerbated hepatic steatosis (31). Thus, the cell-specific metabolic effects of thyroid hormone are largely unknown, and cracking the code requires understanding the deiodinase pathways.

A glimpse into this world is available through the studies in which D2 and D3 expression reciprocally affect energy expenditure in a number of cell and animal models. For example, cAMP-dependent induction of D2 expression during activation of brown adipocytes by cold exposure or high-fat diet enhances local thyroid hormone signaling and energy expenditure, the absence of which prevents normal BAT function (3134). On the other hand, hypoxia-inducible factor-1α-dependent induction of D3 in myocardium and brain during ischemia and hypoxia decreases energy expenditure, supposedly as part of a homeostatic mechanism to slow down cell metabolism in the face of limited O2 supply (19, 35). In fact, D3 reactivation in disease states can be so powerful that it compromises systemic thyroid economy, leading to euthyroid sick syndrome (36). In rare instances, D3-mediated thyroid hormone inactivation is so dramatic that it exceeds the thyroidal synthetic capacity to sustain thyroid economy, leading to consumptive hypothyroidism (37).

D2 expression is the target of a rapidly growing number of molecules that accelerate energy expenditure and metabolic programs in cells and animal models. These include bile acids (38), flavonols (39), and chemical chaperones (40), which in-turn confer protection against diet-induced obesity. Insulin and peroxisome proliferator-activated receptor γ agonists are also bona fide inducers of D2 in skeletal muscle (41). On the other hand, signaling through the D2 pathway is dampened by ER stress (42) and the LXR-RXR pathway (43), the metabolic consequence of which is currently under investigation.

Deiodinases and the Development of Metabolically Relevant Tissues

During vertebrate embryogenesis, developmental signals control the expression interplay between D2 and D3 in metabolic relevant tissues, such as BAT (44), pancreatic islets, and skeletal muscle (15), explaining how “systemic” thyroid hormone can affect local control of tissue embryogenesis.

In the 3-d developmental snapshot during which BAT develops in mice [embryonic day (E)16.5–E18.5], D2 expression is up-regulated about 5-fold and D3 expression drops by 75%. This results in increased local net T3 availability, whereas serum T3 remains unchanged. This rapid enhancement in thyroid hormone signaling is critical for the expression of genes defining BAT identity, i.e. uncoupling protein (UCP)1, peroxisome proliferator-activated receptor gamma co-activator-1α, and Dio2 (44). Notably, these changes in gene expression are observed in utero, without a thermogenic challenge, which highlights the relevance of D2 and its ability to amplify thyroid hormone signaling in a developmental setting. The inactivation of the Dio2 gene as in the D2KO mouse results in a permanent BAT thermogenic defect, compromising thermoregulation and the ability to dissipate excessive calories from diet (31, 32).

The D2 pathway seems to also be critical for skeletal muscle development and function (15). Besides regulating insulin sensitivity in myocytes (41), D2-mediated T3 production is also required for the T3-dependent expression of myogenic factors, such as the myogenic regulatory factor (MyoD), which drive myocyte development. As in BAT, myocytes from D2KO mice have impaired development and function supposedly due to lower intracellular T3 generation. The control of the D2 pathway in myocytes is dependent on the transcription factor FoxO3, which directly binds to the Dio2 promoter, up-regulating D2 expression. The fact that FoxO3 KO myocytes also display impaired cellular development, easily reversed by the addition of exogenous T3, underscores the physiological relevance of the FoxO3/D2 interplay.

The opposite scenario is observed during development of the pancreatic β-cells, with D3 expression keeping thyroid hormone signaling to a minimum, from late embryonic development throughout adulthood (45). The late emergence of D3 expression at E17.5 is restricted to insulin positive cells, indicating a focused role in β-cell but not α-cell development. α-Cell development occurs at a much earlier phase of embryogenesis (by E9.5). As a result of untimely expression of thyroid hormone, D3KO animals exhibit a reduction in total islet area due to decreased β-cells area, insulin content and lower expression of key islet genes involved in glucose sensing, insulin expression, and exocytosis. This is physiologically significant given that adult D3KO animals are glucose intolerant due to impaired glucose-stimulated insulin secretion, without changes in peripheral sensitivity to insulin.

Deiodinases in the Medial Basal Hypothalamus (MBH)

In the central nervous system, D2 is expressed in astrocytes, whereas thyroid hormone receptor and D3 are found in adjacent neurons (46, 47). Thus, glial cell D2 produces T3, which acts in a paracrine fashion to induce thyroid hormone-responsive genes in the nearby neurons (35), a process that is also modulated by D3 activity in the neurons. This paracrine pathway of thyroid hormone action depends on the deiodinases and is thus regulated by signals such as hypoxia, hedgehog signaling, and lipopolysaccharide-induced inflammation, as evidenced both in vitro as well as in rat models of brain ischemia and mouse models of inflammation (48). Therefore, as in other tissues, it is clear that deiodinases function as control points for the regulation of thyroid signaling in the brain.

The neurons in MBH are a target of thyroid hormone, and thus, local D2 and D3 expression can affect thyroid economy and a number of other homeostatic functions (46, 47, 49, 50). Within the MBH, D2 expression is largely restricted to the tanycytes, which are ependymal cells lining the floor and infralateral walls of the third ventricle extending from the rostral tip of the median eminence (ME) to the infundibular recess, surrounding blood vessels in the arcuate nucleus (ARC), and in the ME adjacent to the portal vessels and overlying the tuberoinfundibular sulci (49, 50). Thus, the tanycytes seem to be a major source of T3 to the ARC-ME region of the hypothalamus, likely with important metabolic consequences.

For example, hypothalamic D2 activity in rodents exhibits a circadian rhythmicity with an activity peak at night, which coincides with their peak of metabolic activity (51). At the same time, fasting induces a state of central hypothyroidism that has been linked to an approximately 2-fold up-regulation of D2 expression in the hypothalamus (52) and suppression in TRH/TSH secretion. It has also been suggested that D2 expression in the ARC is localized in glial cells that are in direct opposition to neurons coexpressing neuropeptide Y, Agouti-related peptide, and UCP2 (53). Notably, the fasting-induced increase in D2 activity and local thyroid hormone activation in the ARC is paralleled by an increase in UCP2-dependent mitochondrial uncoupling in neuropeptide Y/Agouti-related peptide expressing neurons. These events were shown to be linked to the increased excitability of these orexigenic neurons and consequent rebound feeding after food deprivation (54).

Deiodinases and the Skeleton

There are strong links emerging between metabolic control and bone and the role of the deiodinases in the skeleton (55). The skeleton is a target of thyroid hormone, which responds by accelerating bone turnover to the extent that there is a net loss of bone mass during systemic thyrotoxicosis (56). In mice, thyroid hormone signaling is kept to a minimum during early bone development due to the high D3 expression (57). Later, during E14.5–E18.5, there is a decrease in D3 and an increase in D2 expression, thus increasing thyroid hormone signaling toward the end of gestation (57). Studies in the developing chicken skeleton indicate that hedgehog signaling mediates the reciprocal control of D2 and D3 expression, transcriptionally increasing Dio3 gene expression and inactivating D2 via induction of WSB-1, the E3 ligase Ub adaptor that ubiquitinates D2 (18, 25). In adult mice, D2 is present in whole-bone extracts, as well as in skeletal cells and differentiated osteoblasts (58), but it is undetectable in chondrocytes and osteoclasts (59). Its absence, as in the D2KO mouse, results in brittle bones due to reduced bone formation, without changes in bone resorption (60). T3 target gene analysis indicates osteoblastic T3 deficiency (60), suggesting that D2-mediated T3 production in osteoblasts is important for maintenance of adult bone mineralization and optimal bone strength.

Conclusion

Thyroid hormone signaling is a local event, with target cells playing a major role through controlled expression of the activating or inactivating deiodinases. Although it is conceivable that plasma T3 plays a metabolic role in some tissues, its relative constancy throughout adult life precludes it from controlling major metabolic pathways. The local role played by the deiodinases in customizing thyroid hormone signaling is the predominant modus operandi through which thyroid hormone exerts its metabolic effects, including in the BAT, β-cell, MBH, bone, and skeletal muscle. Much of this new paradigm of thyroid hormone action was validated through the study of mice with targeted disruption of the deiodinase genes (61). Much remains to be learned while we decipher this code, particularly through the use of a new generation of tissue-specific deiodinase KO animals (62). The consequence of this new way of looking at thyroid hormone action has very significant clinical implications, because serum hormone levels may not be predictive of events that are driving clinical symptoms. This is well illustrated by the series of reports correlating polymorphisms in the three deiodinase genes with a growing number of diseases and clinical conditions in individuals with normal thyroid function tests (63).

Acknowledgments

I thank Dr. Valerie Galton, Dr. Donald St. Germain, and Dr. Arturo Hernandez for graciously sharing different deiodinase KO mouse models and Rafael Arroyo e Drigo, Dr. Tatiana Fonseca, and Dr. Barry Hudson for reviewing the manuscript.

This work was supported in part by National Institute of Diabetes and Digestive and Kidney Diseases Grants DK58538, DK65055, DK77148, and DK7856.

Disclosure Summary: The author has nothing to disclose.

Footnotes

Abbreviations:
ARC
Arcuate nucleus
BAT
brown adipose tissue
D2
type 2 deiodinase
D3
type 3 deiodinase
D2KO
D2 knockout
E
embryonic day
ER
endoplasmic reticulum
Fox
forkhead box
MBH
medial basal hypothalamus
ME
median eminence
Ub
ubiquitin
UCP
uncoupling protein
USP
Ub-specific protease.

References

  • 1. Heuer H, Visser TJ. 2009. Minireview: pathophysiological importance of thyroid hormone transporters. Endocrinology 150:1078–1083 [DOI] [PubMed] [Google Scholar]
  • 2. Cheng SY, Leonard JL, Davis PJ. 2010. Molecular aspects of thyroid hormone actions. Endocr Rev 31:139–170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Bianco AC, Larsen PR. 2005. Cellular and structural biology of the deiodinases. Thyroid 15:777–786 [DOI] [PubMed] [Google Scholar]
  • 4. Baqui M, Botero D, Gereben B, Curcio C, Harney JW, Salvatore D, Sorimachi K, Larsen PR, Bianco AC. 2003. Human type 3 iodothyronine selenodeiodinase is located in the plasma membrane and undergoes rapid internalization to endosomes. J Biol Chem 278:1206–1211 [DOI] [PubMed] [Google Scholar]
  • 5. Baqui MM, Gereben B, Harney JW, Larsen PR, Bianco AC. 2000. Distinct subcellular localization of transiently expressed types 1 and 2 iodothyronine deiodinases as determined by immunofluorescence confocal microscopy. Endocrinology 141:4309–4312 [DOI] [PubMed] [Google Scholar]
  • 6. Curcio-Morelli C, Gereben B, Zavacki AM, Kim BW, Huang S, Harney JW, Larsen PR, Bianco AC. 2003. In vivo dimerization of types 1, 2, and 3 iodothyronine selenodeiodinases. Endocrinology 144:3438–3443 [DOI] [PubMed] [Google Scholar]
  • 7. Callebaut I, Curcio-Morelli C, Mornon JP, Gereben B, Buettner C, Huang S, Castro B, Fonseca TL, Harney JW, Larsen PR, Bianco AC. 2003. The iodothyronine selenodeiodinases are thioredoxin-fold family proteins containing a glycoside hydrolase clan GH-A-like structure. J Biol Chem 278:36887–36896 [DOI] [PubMed] [Google Scholar]
  • 8. Berry MJ, Banu L, Larsen PR. 1991. Type I iodothyronine deiodinase is a selenocysteine-containing enzyme. Nature 349:438–440 [DOI] [PubMed] [Google Scholar]
  • 9. Sagar GD, Gereben B, Callebaut I, Mornon JP, Zeöld A, Curcio-Morelli C, Harney JW, Luongo C, Mulcahey MA, Larsen PR, Huang SA, Bianco AC. 2008. The thyroid hormone-inactivating deiodinase functions as a homodimer. Mol Endocrinol 22:1382–1393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Zeöld A, Pormüller L, Dentice M, Harney JW, Curcio-Morelli C, Tente SM, Bianco AC, Gereben B. 2006. Metabolic instability of type 2 deiodinase is transferable to stable proteins independently of subcellular localization. J Biol Chem 281:31538–31543 [DOI] [PubMed] [Google Scholar]
  • 11. Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, Simonides WS, Zeöld A, Bianco AC. 2008. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocr Rev 29:898–938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gereben B, Zeöld A, Dentice M, Salvatore D, Bianco AC. 2008. Activation and inactivation of thyroid hormone by deiodinases: local action with general consequences. Cell Mol Life Sci 65:570–590 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Bartha T, Kim SW, Salvatore D, Gereben B, Tu HM, Harney JW, Rudas P, Larsen PR. 2000. Characterization of the 5′-flanking and 5′-untranslated regions of the cyclic adenosine 3′,5′-monophosphate-responsive human type 2 iodothyronine deiodinase gene. Endocrinology 141:229–237 [DOI] [PubMed] [Google Scholar]
  • 14. Fekete C, Gereben B, Doleschall M, Harney JW, Dora JM, Bianco AC, Sarkar S, Liposits Z, Rand W, Emerson C, Kacskovics I, Larsen PR, Lechan RM. 2004. Lipopolysaccharide induces type 2 iodothyronine deiodinase in the mediobasal hypothalamus: implications for the nonthyroidal illness syndrome. Endocrinology 145:1649–1655 [DOI] [PubMed] [Google Scholar]
  • 15. Dentice M, Marsili A, Ambrosio R, Guardiola O, Sibilio A, Paik JH, Minchiotti G, DePinho RA, Fenzi G, Larsen PR, Salvatore D. 2010. The FoxO3/type 2 deiodinase pathway is required for normal mouse myogenesis and muscle regeneration. J Clin Invest 120:4021–4030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Pallud S, Ramaugé M, Gavaret JM, Lennon AM, Munsch N, St Germain DL, Pierre M, Courtin F. 1999. Regulation of type 3 iodothyronine deiodinase expression in cultured rat astrocytes: role of the Erk cascade. Endocrinology 140:2917–2923 [DOI] [PubMed] [Google Scholar]
  • 17. Huang SA, Mulcahey MA, Crescenzi A, Chung M, Kim BW, Barnes C, Kuijt W, Turano H, Harney J, Larsen PR. 2005. TGF-B promotes inactivation of extracellular thyroid hormones via transcriptional stimulation of type 3 iodothyronine deiodinase. Mol Endocrinol 19:3126–3136 [DOI] [PubMed] [Google Scholar]
  • 18. Dentice M, Luongo C, Huang S, Ambrosio R, Elefante A, Mirebeau-Prunier D, Zavacki AM, Fenzi G, Grachtchouk M, Hutchin M, Dlugosz AA, Bianco AC, Missero C, Larsen PR, Salvatore D. 2007. Sonic hedgehog-induced type 3 deiodinase blocks thyroid hormone action enhancing proliferation of normal and malignant keratinocytes. Proc Natl Acad Sci USA 104:14466–14471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Simonides WS, Mulcahey MA, Redout EM, Muller A, Zuidwijk MJ, Visser TJ, Wassen FW, Crescenzi A, da-Silva WS, Harney J, Engel FB, Obregon MJ, Larsen PR, Bianco AC, Huang SA. 2008. Hypoxia-inducible factor induces local thyroid hormone inactivation during hypoxic-ischemic disease in rats. J Clin Invest 118:975–983 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Sagar GD, Gereben B, Callebaut I, Mornon JP, Zeöld A, da Silva WS, Luongo C, Dentice M, Tente SM, Freitas BC, Harney JW, Zavacki AM, Bianco AC. 2007. Ubiquitination-induced conformational change within the deiodinase dimer is a switch regulating enzyme activity. Mol Cell Biol 27:4774–4783 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Curcio-Morelli C, Zavacki AM, Christofollete M, Gereben B, de Freitas BC, Harney JW, Li Z, Wu G, Bianco AC. 2003. Deubiquitination of type 2 iodothyronine deiodinase by von Hippel-Lindau protein-interacting deubiquitinating enzymes regulates thyroid hormone activation. J Clin Invest 112:189–196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Gereben B, Goncalves C, Harney JW, Larsen PR, Bianco AC. 2000. Selective proteolysis of human type 2 deiodinase: a novel ubiquitin-proteasomal mediated mechanism for regulation of hormone activation. Mol Endocrinol 14:1697–1708 [DOI] [PubMed] [Google Scholar]
  • 23. Botero D, Gereben B, Goncalves C, De Jesus LA, Harney JW, Bianco AC. 2002. Ubc6p and Ubc7p are required for normal and substrate-induced endoplasmic reticulum-associated degradation of the human selenoprotein type 2 iodothyronine monodeiodinase. Mol Endocrinol 16:1999–2007 [DOI] [PubMed] [Google Scholar]
  • 24. Kim BW, Zavacki AM, Curcio-Morelli C, Dentice M, Harney JW, Larsen PR, Bianco AC. 2003. Endoplasmic reticulum-associated degradation of the human type 2 iodothyronine deiodinase (D2) is mediated via an association between mammalian UBC7 and the carboxyl region of D2. Mol Endocrinol 17:2603–2612 [DOI] [PubMed] [Google Scholar]
  • 25. Dentice M, Bandyopadhyay A, Gereben B, Callebaut I, Christoffolete MA, Kim BW, Nissim S, Mornon JP, Zavacki AM, Zeöld A, Capelo LP, Curcio-Morelli C, Ribeiro R, Harney JW, Tabin CJ, Bianco AC. 2005. The Hedgehog-inducible ubiquitin ligase subunit WSB-1 modulates thyroid hormone activation and PTHrP secretion in the developing growth plate. Nat Cell Biol 7:698–705 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Zavacki AM, Arrojo E, Drigo R, Freitas BC, Chung M, Harney JW, Egri P, Wittmann G, Fekete C, Gereben B, Bianco AC. 2009. The E3 ubiquitin ligase TEB4 mediates degradation of type 2 iodothyronine deiodinase. Mol Cell Biol 29:5339–5347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Andersen S, Bruun NH, Pedersen KM, Laurberg P. 2003. Biologic variation is important for interpretation of thyroid function tests. Thyroid 13:1069–1078 [DOI] [PubMed] [Google Scholar]
  • 28. Bianco AC, Maia AL, da Silva WS, Christoffolete MA. 2005. Adaptive activation of thyroid hormone and energy expenditure. Biosci Rep 25:191–208 [DOI] [PubMed] [Google Scholar]
  • 29. Silva JE. 2000. Catecholamines and the sympathoadrenal system in thyrotoxicosis. In: Braverman LE, Utiger RD. eds. Werner and Ingbar's the thyroid: a fundamental and clinical text. Philadelphia: Lippincott, Willians & Wilkins; 642–651 [Google Scholar]
  • 30. Silva JE. 2000. Catecholamines and the sympathoadrenal system in hypothyroidism. In: Braverman LE, Utiger RD. eds. Werner and Ingbar's the thyroid: a fundamental and clinical text. Philadelphia: Lippincott, Willians & Wilkins; 820–823 [Google Scholar]
  • 31. Castillo M, Hall JA, Correa-Medina M, Ueta C, Won Kang H, Cohen DE, Bianco AC. 2011. Disruption of thyroid hormone activation in type 2 deiodinase knockout mice causes obesity with glucose intolerance and liver steatosis only at thermoneutrality. Diabetes 60:1082–1089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. de Jesus LA, Carvalho SD, Ribeiro MO, Schneider M, Kim SW, Harney JW, Larsen PR, Bianco AC. 2001. The type 2 iodothyronine deiodinase is essential for adaptive thermogenesis in brown adipose tissue. J Clin Invest 108:1379–1385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Bianco AC, Carvalho SD, Carvalho CR, Rabelo R, Moriscot AS. 1998. Thyroxine 5′-deiodination mediates norepinephrine-induced lipogenesis in dispersed brown adipocytes. Endocrinology 139:571–578 [DOI] [PubMed] [Google Scholar]
  • 34. Bianco AC, Silva JE. 1987. Intracellular conversion of thyroxine to triiodothyronine is required for the optimal thermogenic function of brown adipose tissue. J Clin Invest 79:295–300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Freitas BC, Gereben B, Castillo M, Kalló I, Zeöld A, Egri P, Liposits Z, Zavacki AM, Maciel RM, Jo S, Singru P, Sanchez E, Lechan RM, Bianco AC. 2010. Paracrine signaling by glial cell-derived triiodothyronine activates neuronal gene expression in the rodent brain and human cells. J Clin Invest 120:2206–2217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Huang SA, Bianco AC. 2008. Reawakened interest in type III iodothyronine deiodinase in critical illness and injury. Nat Clin Pract Endocrinol Metab 4:148–155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Huang SA, Tu HM, Harney JW, Venihaki M, Butte AJ, Kozakewich HP, Fishman SJ, Larsen PR. 2000. Severe hypothyroidism caused by type 3 iodothyronine deiodinase in infantile hemangiomas. New Engl J Med 343:185–189 [DOI] [PubMed] [Google Scholar]
  • 38. Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, Messaddeq N, Harney JW, Ezaki O, Kodama T, Schoonjans K, Bianco AC, Auwerx J. 2006. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439:484–489 [DOI] [PubMed] [Google Scholar]
  • 39. da-Silva WS, Harney JW, Kim BW, Li J, Bianco SD, Crescenzi A, Christoffolete MA, Huang SA, Bianco AC. 2007. The small polyphenolic molecule kaempferol increases cellular energy expenditure and thyroid hormone activation. Diabetes 56:767–776 [DOI] [PubMed] [Google Scholar]
  • 40. da-Silva WS, Ribich S, Arrojo e Drigo R, Castillo M, Patti ME, Bianco AC. 2011. The chemical chaperones tauroursodeoxycholic and 4-phenylbutyric acid accelerate thyroid hormone activation and energy expenditure. FEBS Lett 585:539–544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Grozovsky R, Ribich S, Rosene ML, Mulcahey MA, Huang SA, Patti ME, Bianco AC, Kim BW. 2009. Type 2 deiodinase expression is induced by peroxisomal proliferator-activated receptor-γ agonists in skeletal myocytes. Endocrinology 150:1976–1983 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Arrojo e Drigo R, Fonseca TL, Castillo M, Simovic G, Gereben G, Bianco AC. Endoplasmatic reticulum stress and chemical chaperones regulate type 2 deiodinase and thyroid hormone signaling. International Thyroid Congress, Paris, 2010 [Google Scholar]
  • 43. Christoffolete MA, Doleschall M, Egri P, Liposits Z, Zavacki AM, Bianco AC, Gereben B. 2010. Regulation of thyroid hormone activation via the liver X-receptor/retinoid X-receptor pathway. J Endocrinol 205:179–186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Hall JA, Ribich S, Cristoffolete MA, Simovic G, Correa-Medina M, Patti ME, Bianco AC. 2010. Absence of thyroid hormone activation during development underlies a permanent defect in adaptive thermogenesis. Endocrinology 151:4573–4582 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Correa M, Molina J, Gadea Y, Gereben B, Fachado A, Pileggi A, Hernandez A, Edlund H, Bianco AC. Type 3 deiodinase in pancreatic B-cell is critical for glucose homeostasis. International Thyroid Congress, Paris, 2010 [Google Scholar]
  • 46. Lechan RM, Fekete C. 2005. Role of thyroid hormone deiodination in the hypothalamus. Thyroid 15:883–897 [DOI] [PubMed] [Google Scholar]
  • 47. Hollenberg AN. 2008. The role of the thyrotropin-releasing hormone (TRH) neuron as a metabolic sensor. Thyroid 18:131–139 [DOI] [PubMed] [Google Scholar]
  • 48. Fekete C, Sarkar S, Christoffolete MA, Emerson CH, Bianco AC, Lechan RM. 2005. Bacterial lipopolysaccharide (LPS)-induced type 2 iodothyronine deiodinase (D2) activation in the mediobasal hypothalamus (MBH) is independent of the LPS-induced fall in serum thyroid hormone levels. Brain Res 1056:97–99 [DOI] [PubMed] [Google Scholar]
  • 49. Tu HM, Kim SW, Salvatore D, Bartha T, Legradi G, Larsen PR, Lechan RM. 1997. Regional distribution of type 2 thyroxine deiodinase messenger ribonucleic acid in rat hypothalamus and pituitary and its regulation by thyroid hormone. Endocrinology 138:3359–3368 [DOI] [PubMed] [Google Scholar]
  • 50. Guadaño-Ferraz A, Obregón MJ, St Germain DL, Bernal J. 1997. The type 2 iodothyronine deiodinase is expressed primarily in glial cells in the neonatal rat brain. Proc Natl Acad Sci USA 94:10391–10396 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Campos-Barros A, Musa A, Flechner A, Hessenius C, Gaio U, Meinhold H, Baumgartner A. 1997. Evidence for circadian variations of thyroid hormone concentrations and type II 5′-iodothyronine deiodinase activity in the rat central nervous system. J Neurochem 68:795–803 [DOI] [PubMed] [Google Scholar]
  • 52. Diano S, Naftolin F, Goglia F, Horvath TL. 1998. Fasting-induced increase in type II iodothyronine deiodinase activity and messenger ribonucleic acid levels is not reversed by thyroxine in the rat hypothalamus. Endocrinology 139:2879–2884 [DOI] [PubMed] [Google Scholar]
  • 53. Fekete C, Sarkar S, Rand WM, Harney JW, Emerson CH, Bianco AC, Beck-Sickinger A, Lechan RM. 2002. Neuropeptide Y1 and Y5 receptors mediate the effects of neuropeptide Y on the hypothalamic-pituitary-thyroid axis. Endocrinology 143:4513–4519 [DOI] [PubMed] [Google Scholar]
  • 54. Coppola A, Liu ZW, Andrews ZB, Paradis E, Roy MC, Friedman JM, Ricquier D, Richard D, Horvath TL, Gao XB, Diano S. 2007. A central thermogenic-like mechanism in feeding regulation: an interplay between arcuate nucleus T3 and UCP2. Cell Metab 5:21–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Williams GR, Bassett JH. 2011. Local control of thyroid hormone action - role of type 2 deiodinase. J Endocrinol 209:261–272 [DOI] [PubMed] [Google Scholar]
  • 56. Murphy E, Williams GR. 2004. The thyroid and the skeleton. Clin Endocrinol 61:285–298 [DOI] [PubMed] [Google Scholar]
  • 57. Capelo LP, Beber EH, Huang SA, Zorn TM, Bianco AC, Gouveia CH. 2008. Deiodinase-mediated thyroid hormone inactivation minimizes thyroid hormone signaling in the early development of fetal skeleton. Bone 43:921–930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Gouveia CH, Christoffolete MA, Zaitune CR, Dora JM, Harney JW, Maia AL, Bianco AC. 2005. Type 2 iodothyronine selenodeiodinase is expressed throughout the mouse skeleton and in the MC3T3-E1 mouse osteoblastic cell line during differentiation. Endocrinology 146:195–200 [DOI] [PubMed] [Google Scholar]
  • 59. Williams AJ, Robson H, Kester MH, van Leeuwen JP, Shalet SM, Visser TJ, Williams GR. 2008. Iodothyronine deiodinase enzyme activities in bone. Bone 43:126–134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Bassett JH, Boyde A, Howell PG, Bassett RH, Galliford TM, Archanco M, Evans H, Lawson MA, Croucher P, St Germain DL, Galton VA, Williams GR. 2010. Optimal bone strength and mineralization requires the type 2 iodothyronine deiodinase in osteoblasts. Proc Natl Acad Sci USA 107:7604–7609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. St Germain DL, Hernandez A, Schneider MJ, Galton VA. 2005. Insights into the role of deiodinases from studies of genetically modified animals. Thyroid 15:905–916 [DOI] [PubMed] [Google Scholar]
  • 62. Fonseca TL, Ueta CB, Campos MPO, Medina MC, Rosene M, Gereben B, Bianco AC. Tissue-specific deletion of the type 2 deiodinase gene identifies a role for pituitary D2 in the TSH feedback mechanism. International Thyroid Congress, Paris, 2010 [Google Scholar]
  • 63. Peeters RP, van der Deure WM, Visser TJ. 2006. Genetic variation in thyroid hormone pathway genes; polymorphisms in the TSH receptor and the iodothyronine deiodinases. Eur J Endocrinol 155:655–662 [DOI] [PubMed] [Google Scholar]

Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES